1
|
Szwed-Georgiou A, Płociński P, Kupikowska-Stobba B, Urbaniak MM, Rusek-Wala P, Szustakiewicz K, Piszko P, Krupa A, Biernat M, Gazińska M, Kasprzak M, Nawrotek K, Mira NP, Rudnicka K. Bioactive Materials for Bone Regeneration: Biomolecules and Delivery Systems. ACS Biomater Sci Eng 2023; 9:5222-5254. [PMID: 37585562 PMCID: PMC10498424 DOI: 10.1021/acsbiomaterials.3c00609] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/31/2023] [Indexed: 08/18/2023]
Abstract
Novel tissue regeneration strategies are constantly being developed worldwide. Research on bone regeneration is noteworthy, as many promising new approaches have been documented with novel strategies currently under investigation. Innovative biomaterials that allow the coordinated and well-controlled repair of bone fractures and bone loss are being designed to reduce the need for autologous or allogeneic bone grafts eventually. The current engineering technologies permit the construction of synthetic, complex, biomimetic biomaterials with properties nearly as good as those of natural bone with good biocompatibility. To ensure that all these requirements meet, bioactive molecules are coupled to structural scaffolding constituents to form a final product with the desired physical, chemical, and biological properties. Bioactive molecules that have been used to promote bone regeneration include protein growth factors, peptides, amino acids, hormones, lipids, and flavonoids. Various strategies have been adapted to investigate the coupling of bioactive molecules with scaffolding materials to sustain activity and allow controlled release. The current manuscript is a thorough survey of the strategies that have been exploited for the delivery of biomolecules for bone regeneration purposes, from choosing the bioactive molecule to selecting the optimal strategy to synthesize the scaffold and assessing the advantages and disadvantages of various delivery strategies.
Collapse
Affiliation(s)
- Aleksandra Szwed-Georgiou
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Przemysław Płociński
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Barbara Kupikowska-Stobba
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Mateusz M. Urbaniak
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
- The
Bio-Med-Chem Doctoral School, University of Lodz and Lodz Institutes
of the Polish Academy of Sciences, University
of Lodz, Lodz 90-237, Poland
| | - Paulina Rusek-Wala
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
- The
Bio-Med-Chem Doctoral School, University of Lodz and Lodz Institutes
of the Polish Academy of Sciences, University
of Lodz, Lodz 90-237, Poland
| | - Konrad Szustakiewicz
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Paweł Piszko
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Agnieszka Krupa
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Monika Biernat
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Małgorzata Gazińska
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Mirosław Kasprzak
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Katarzyna Nawrotek
- Faculty
of Process and Environmental Engineering, Lodz University of Technology, Lodz 90-924, Poland
| | - Nuno Pereira Mira
- iBB-Institute
for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de
Lisboa, Lisboa 1049-001, Portugal
- Associate
Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior
Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
- Instituto
Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Karolina Rudnicka
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| |
Collapse
|
2
|
Akhmetshina A, Kratky D, Rendina-Ruedy E. Influence of Cholesterol on the Regulation of Osteoblast Function. Metabolites 2023; 13:metabo13040578. [PMID: 37110236 PMCID: PMC10143138 DOI: 10.3390/metabo13040578] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/11/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Bone is a dynamic tissue composed of cells, an extracellular matrix, and mineralized portion. Osteoblasts are responsible for proper bone formation and remodeling, and function. These processes are endergonic and require cellular energy in the form of adenosine triphosphate (ATP), which is derived from various sources such as glucose, fatty acids, and amino acids. However, other lipids such as cholesterol have also been found to play a critical role in bone homeostasis and can also contribute to the overall bioenergetic capacity of osteoblasts. In addition, several epidemiological studies have found a link between elevated cholesterol, cardiovascular disease, an enhanced risk of osteoporosis, and increased bone metastasis in cancer patients. This review focuses on how cholesterol, its derivatives, and cholesterol-lowering medications (statins) regulate osteoblast function and bone formation. It also highlights the molecular mechanisms underlying the cholesterol-osteoblast crosstalk.
Collapse
Affiliation(s)
- Alena Akhmetshina
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Elizabeth Rendina-Ruedy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
3
|
Zhang Y, Zhou J, Wu JL, Ma JC, Wang H, Wen J, Huang S, Lee M, Bai X, Cui ZK. Intrinsic antibacterial and osteoinductive sterosomes promote infected bone healing. J Control Release 2023; 354:713-725. [PMID: 36702258 DOI: 10.1016/j.jconrel.2023.01.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 01/08/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023]
Abstract
Open fractures and internal fixation implants are often accompanied by bacterial infection, leading to osteomyelitis, characterized by intractable bone infection and sequestrum formation, and can result in lifelong disability or fatal sepsis. As common clinical treatment strategies, high-dose antibiotic application and autologous bone transplantation face the risk of recurrence and donor site injury. Herein, we designed and prepared a novel drug delivery system by rational selection of the antibacterial single-chain amphiphile (cetylpyridinium chloride, CPC) and osteoinductive sterol (20S-hydroxycholesterol, Oxy) to formulate CPC/Oxy sterosomes. We demonstrate their excellent biocompatibility and antibacterial ability through 2D and 3D settings in vitro. In addition, the osteogenic differentiation of bone marrow mesenchymal stem cells was investigated in cell monolayers and a hydrogel environment. Moreover, a rat infected critical-sized calvarial defect model was employed to illustrate the effects of antibacterial and osteogenic CPC/Oxy sterosomes in vivo. Our results showed that CPC/Oxy sterosomes not only exterminated bacterial infections, but also enhanced calvarial healing without additional antibiotics, bone formation promoters or exogenous cells. This research provides a promising and effective multifunctional sterosomal platform for the treatment of infected bone defects, with the potential to be combined with therapeutic genes, and small molecule drugs.
Collapse
Affiliation(s)
- Yiqing Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jie Zhou
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiao-Lan Wu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jian-Chao Ma
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hui Wang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Jing Wen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shen Huang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Min Lee
- Division of Advanced Prosthodontics, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhong-Kai Cui
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Spine Surgery, Ganzhou People's Hospital, Ganzhou 342800, China.
| |
Collapse
|
4
|
Oxysterols are potential physiological regulators of ageing. Ageing Res Rev 2022; 77:101615. [PMID: 35351610 DOI: 10.1016/j.arr.2022.101615] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/18/2022] [Accepted: 03/24/2022] [Indexed: 12/24/2022]
Abstract
Delaying and even reversing ageing is a major public health challenge with a tremendous potential to postpone a plethora of diseases including cancer, metabolic syndromes and neurodegenerative disorders. A better understanding of ageing as well as the development of innovative anti-ageing strategies are therefore an increasingly important field of research. Several biological processes including inflammation, proteostasis, epigenetic, oxidative stress, stem cell exhaustion, senescence and stress adaptive response have been reported for their key role in ageing. In this review, we describe the relationships that have been established between cholesterol homeostasis, in particular at the level of oxysterols, and ageing. Initially considered as harmful pro-inflammatory and cytotoxic metabolites, oxysterols are currently emerging as an expanding family of fine regulators of various biological processes involved in ageing. Indeed, depending of their chemical structure and their concentration, oxysterols exhibit deleterious or beneficial effects on inflammation, oxidative stress and cell survival. In addition, stem cell differentiation, epigenetics, cellular senescence and proteostasis are also modulated by oxysterols. Altogether, these data support the fact that ageing is influenced by an oxysterol profile. Further studies are thus required to explore more deeply the impact of the "oxysterome" on ageing and therefore this cholesterol metabolic pathway constitutes a promising target for future anti-ageing interventions.
Collapse
|
5
|
New Function of Cholesterol Oxidation Products Involved in Osteoporosis Pathogenesis. Int J Mol Sci 2022; 23:ijms23042020. [PMID: 35216140 PMCID: PMC8876989 DOI: 10.3390/ijms23042020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/05/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis (OP) is a systemic bone disease characterized by decreased bone strength, microarchitectural changes in bone tissues, and increased risk of fracture. Its occurrence is closely related to various factors such as aging, genetic factors, living habits, and nutritional deficiencies as well as the disturbance of bone homeostasis. The dysregulation of bone metabolism is regarded as one of the key influencing factors causing OP. Cholesterol oxidation products (COPs) are important compounds in the maintenance of bone metabolic homeostasis by participating in several important biological processes such as the differentiation of mesenchymal stem cells, bone formation in osteoblasts, and bone resorption in osteoclasts. The effects of specific COPs on mesenchymal stem cells are mainly manifested by promoting osteoblast genesis and inhibiting adipocyte genesis. This review aims to elucidate the biological roles of COPs in OP development, starting from the molecular mechanisms of OP, pointing out opportunities and challenges in current research, and providing new ideas and perspectives for further studies of OP pathogenesis.
Collapse
|
6
|
de Freitas FA, Levy D, Zarrouk A, Lizard G, Bydlowski SP. Impact of Oxysterols on Cell Death, Proliferation, and Differentiation Induction: Current Status. Cells 2021; 10:cells10092301. [PMID: 34571949 PMCID: PMC8468221 DOI: 10.3390/cells10092301] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/16/2022] Open
Abstract
Oxysterols are oxidized derivatives of cholesterol produced by enzymatic activity or non-enzymatic pathways (auto-oxidation). The oxidation processes lead to the synthesis of about 60 different oxysterols. Several oxysterols have physiological, pathophysiological, and pharmacological activities. The effects of oxysterols on cell death processes, especially apoptosis, autophagy, necrosis, and oxiapoptophagy, as well as their action on cell proliferation, are reviewed here. These effects, also observed in several cancer cell lines, could potentially be useful in cancer treatment. The effects of oxysterols on cell differentiation are also described. Among them, the properties of stimulating the osteogenic differentiation of mesenchymal stem cells while inhibiting adipogenic differentiation may be useful in regenerative medicine.
Collapse
Affiliation(s)
- Fábio Alessandro de Freitas
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-900, Brazil (D.L.)
| | - Débora Levy
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-900, Brazil (D.L.)
| | - Amira Zarrouk
- Faculty of Medicine, University of Monastir, LR12ES05, Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Monastir, Tunisia & Faculty of Medicine, University of Sousse, Sousse 5000, Tunisia;
| | - Gérard Lizard
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA, University of Bourgogne Franche-Comté, Institut National de la Santé et de la Recherche Médicale—Inserm, 7270 Dijon, France;
| | - Sérgio Paulo Bydlowski
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-900, Brazil (D.L.)
- National Institute of Science and Technology in Regenerative Medicine (INCT-Regenera), CNPq, Rio de Janeiro 21941-902, Brazil
- Correspondence:
| |
Collapse
|
7
|
Abstract
Mesenchymal stem cells have the ability to differentiate into several cell types when exposed to determined substances, including oxysterols. Oxysterols are cholesterol products derived from its auto-oxidation by reactive species or from enzymatic action. They are present in the body in low quantities under physiological conditions and exhibit several physiological and pharmacological actions according to both the types of oxysterol and tissue. Some of them are cytotoxic while others have been shown to promote cell differentiation through the action on several different receptors, such as nuclear LXR receptors and Smoothened receptor ligands. Here, we review the main pathways by which oxysterols have been associated with cell differentiation and death of mesenchymal stem cells.
Collapse
|
8
|
Goel D, Vohora D. Liver X receptors and skeleton: Current state-of-knowledge. Bone 2021; 144:115807. [PMID: 33333244 DOI: 10.1016/j.bone.2020.115807] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/26/2020] [Accepted: 12/11/2020] [Indexed: 12/25/2022]
Abstract
The liver X receptors (LXR) is a nuclear receptor that acts as a prominent regulator of lipid homeostasis and inflammatory response. Its therapeutic effectiveness against various diseases like Alzheimer's disease and atherosclerosis has been investigated in detail. Emerging pieces of evidence now reveal that LXR is also a crucial modulator of bone remodeling. However, the molecular mechanisms underlying the pharmacological actions of LXR on the skeleton and its role in osteoporosis are poorly understood. Therefore, in the current review, we highlight LXR and its actions through different molecular pathways modulating skeletal homeostasis. The studies described in this review propound that LXR in association with estrogen, PTH, PPARγ, RXR hedgehog, and canonical Wnt signaling regulates osteoclastogenesis and bone resorption. It regulates RANKL-induced expression of c-Fos, NFATc1, and NF-κB involved in osteoclast differentiation. Additionally, several studies suggest suppression of RANKL-induced osteoclast differentiation by synthetic LXR ligands. Given the significance of modulation of LXR in various physiological and pathological settings, our findings indicate that therapeutic targeting of LXR might potentially prevent or treat osteoporosis and improve bone quality.
Collapse
Affiliation(s)
- Divya Goel
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi 110062, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
9
|
Samadi A, Sabuncuoglu S, Samadi M, Isikhan SY, Chirumbolo S, Peana M, Lay I, Yalcinkaya A, Bjørklund G. A Comprehensive Review on Oxysterols and Related Diseases. Curr Med Chem 2021; 28:110-136. [PMID: 32175830 DOI: 10.2174/0929867327666200316142659] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/09/2019] [Accepted: 11/10/2019] [Indexed: 11/22/2022]
Abstract
The present review aims to provide a complete and comprehensive summary of current literature relevant to oxysterols and related diseases. Oxidation of cholesterol leads to the formation of a large number of oxidized products, generally known as oxysterols. They are intermediates in the biosynthesis of bile acids, steroid hormones, and 1,25- dihydroxyvitamin D3. Although oxysterols are considered as metabolic intermediates, there is a growing body of evidence that many of them are bioactive, and their absence or excess may be part of the cause of a disease phenotype. These compounds derive from either enzymatic or non-enzymatic oxidation of cholesterol. This study provides comprehensive information about the structures, formation, and types of oxysterols even when involved in certain disease states, focusing on their effects on metabolism and linkages with these diseases. The role of specific oxysterols as mediators in various disorders, such as degenerative (age-related) and cancer-related disorders, has now become clearer. Oxysterol levels may be employed as suitable markers for the diagnosis of specific diseases or in predicting the incidence rate of diseases, such as diabetes mellitus, Alzheimer's disease, multiple sclerosis, osteoporosis, lung cancer, breast cancer, and infertility. However, further investigations may be required to confirm these mentioned possibilities.
Collapse
Affiliation(s)
- Afshin Samadi
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
| | - Suna Sabuncuoglu
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Mahshid Samadi
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Selen Yilmaz Isikhan
- Vocational Higher School of Social Sciences, Hacettepe University, Ankara, Turkey
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Massimiliano Peana
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | - Incilay Lay
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ahmet Yalcinkaya
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| |
Collapse
|
10
|
Tompkins YH, Su S, Velleman SG, Kim WK. Effects of 20(S)-hydroxycholesterol on satellite cell proliferation and differentiation of broilers. Poult Sci 2021; 100:474-481. [PMID: 33518099 PMCID: PMC7858162 DOI: 10.1016/j.psj.2020.10.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/14/2020] [Accepted: 10/19/2020] [Indexed: 01/30/2023] Open
Abstract
In the modern poultry industry, with increasing product demand, muscle growth rate and meat yield in chickens have tremendously changed. Understanding the regulation of muscle development is important to maintain efficient growth and development in meat-type chickens. 20(S)-hydroxycholesterol (20S) is known as one of the naturally occurring osteogenic cholesterol derivatives due to its ability to induce osteogenic differentiation; however, no studies have evaluated myogenic response to 20S in chicken muscle cells. To determine the use of 20S in vitro for the proliferation and differentiation of chicken satellite cells, satellite cells were isolated from pectoralis major muscle of 4-week-old Ross 708 male chickens and subjected to 0.25, 0.5, and 1.0 μmol of 20S during their proliferation and differentiation stages. Cell proliferation and differentiation were measured every 24 h for 72 h by determining DNA concentration, the activity of creatine kinase, and the expressions of myogenic regulatory transcription factors. Together these results suggested that a lower concentration of 20S did not affect myogenesis but a high concentration of 1.0 μmol 20S can negatively affect proliferation and differentiation in chicken satellite cells.
Collapse
Affiliation(s)
- Yuguo H Tompkins
- Department of Poultry Science, University of Georgia, Athens, USA
| | - Shengchen Su
- Department of Poultry Science, University of Georgia, Athens, USA
| | - Sandra G Velleman
- The Ohio State University, Ohio Agricultural Research and Development Center, Wooster, USA
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, USA.
| |
Collapse
|
11
|
Effect of 20(S)-Hydroxycholesterol on Multilineage Differentiation of Mesenchymal Stem Cells Isolated from Compact Bones in Chicken. Genes (Basel) 2020; 11:genes11111360. [PMID: 33213081 PMCID: PMC7698591 DOI: 10.3390/genes11111360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/17/2022] Open
Abstract
Bone health and body weight gain have significant economic and welfare importance in the poultry industry. Mesenchymal stem cells (MSCs) are common progenitors of different cell lineages such as osteoblasts, adipocytes, and myocytes. Specific oxysterols have shown to be pro-osteogenic and anti-adipogenic in mouse and human MSCs. To determine the effect of 20(S)-hydroxycholesterol (20S) on osteogenic, adipogenic, and myogenic differentiation in chicken, mesenchymal stem cells isolated from compact bones of broiler chickens (cBMSCs) were subjected to various doses of 20S, and markers of lineage-specific mRNA were analyzed using real-time PCR and cell cytochemistry. Further studies were conducted to evaluate the molecular mechanisms involved in lineage-specific differentiation pathways. Like human and mouse MSCs, 20S oxysterol expressed pro-osteogenic, pro-myogenic, and anti-adipogenic differentiation potential in cBMSCs. Moreover, 20(S)-Hydroxycholesterol induced markers of osteogenic genes and myogenic regulatory factors when exposed to cBMSCs treated with their specific medium. In contrast, 20S oxysterol suppressed expression of adipogenic marker genes when exposed to cBMSCs treated with OA, an adipogenic precursor of cBMSCs. To elucidate the molecular mechanism by which 20S exerts its differentiation potential in all three lineages, we focused on the hedgehog signaling pathway. The hedgehog inhibitor, cyclopamine, completely reversed the effect of 20S induced expression of osteogenic and anti-adipogenic mRNA. However, there was no change in the mRNA expression of myogenic genes. The results showed that 20S oxysterol promotes osteogenic and myogenic differentiation and decreases adipocyte differentiation of cBMSCs. This study also showed that the induction of osteogenesis and adipogenesis inhibition in cBMSCs by 20S is mediated through the hedgehog signaling mechanism. The results indicated that 20(S) could play an important role in the differentiation of chicken-derived MSCs and provided the theory basis on developing an intervention strategy to regulate skeletal, myogenic, and adipogenic differentiation in chicken, which will contribute to improving chicken bone health and meat quality. The current results provide the rationale for the further study of regulatory mechanisms of bioactive molecules on the differentiation of MSCs in chicken, which can help to address skeletal health problems in poultry.
Collapse
|
12
|
Cottrill E, Lazzari J, Pennington Z, Ehresman J, Schilling A, Dirckx N, Theodore N, Sciubba D, Witham T. Oxysterols as promising small molecules for bone tissue engineering: Systematic review. World J Orthop 2020; 11:328-344. [PMID: 32908817 PMCID: PMC7453739 DOI: 10.5312/wjo.v11.i7.328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/08/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bone tissue engineering is an area of continued interest within orthopaedic surgery, as it promises to create implantable bone substitute materials that obviate the need for autologous bone graft. Recently, oxysterols – oxygenated derivatives of cholesterol – have been proposed as a novel class of osteoinductive small molecules for bone tissue engineering. Here, we present the first systematic review of the in vivo evidence describing the potential therapeutic utility of oxysterols for bone tissue engineering.
AIM To systematically review the available literature examining the effect of oxysterols on in vivo bone formation.
METHODS We conducted a systematic review of the literature following PRISMA guidelines. Using the PubMed/MEDLINE, Embase, and Web of Science databases, we queried all publications in the English-language literature investigating the effect of oxysterols on in vivo bone formation. Articles were screened for eligibility using PICOS criteria and assessed for potential bias using an expanded version of the SYRCLE Risk of Bias assessment tool. All full-text articles examining the effect of oxysterols on in vivo bone formation were included. Extracted data included: Animal species, surgical/defect model, description of therapeutic and control treatments, and method for assessing bone growth. Primary outcome was fusion rate for spinal fusion models and percent bone regeneration for critical-sized defect models. Data were tabulated and described by both surgical/defect model and oxysterol employed. Additionally, data from all included studies were aggregated to posit the mechanism by which oxysterols may mediate in vivo bone formation.
RESULTS Our search identified 267 unique articles, of which 27 underwent full-text review. Thirteen studies (all preclinical) met our inclusion/exclusion criteria. Of the 13 included studies, 5 employed spinal fusion models, 2 employed critical-sized alveolar defect models, and 6 employed critical-sized calvarial defect models. Based upon SYRCLE criteria, the included studies were found to possess an overall “unclear risk of bias”; 54% of studies reported treatment randomization and 38% reported blinding at any level. Overall, seven unique oxysterols were evaluated: 20(S)-hydroxycholesterol, 22(R)-hydroxycholesterol, 22(S)-hydroxycholesterol, Oxy4/Oxy34, Oxy18, Oxy21/Oxy133, and Oxy49. All had statistically significant in vivo osteoinductive properties, with Oxy4/Oxy34, Oxy21/Oxy133, and Oxy49 showing a dose-dependent effect in some cases. In the eight studies that directly compared oxysterols to rhBMP-2-treated animals, similar rates of bone growth occurred in the two groups. Biochemical investigation of these effects suggests that they may be primarily mediated by direct activation of Smoothened in the Hedgehog signaling pathway.
CONCLUSION Present preclinical evidence suggests oxysterols significantly augment in vivo bone formation. However, clinical trials are necessary to determine which have the greatest therapeutic potential for orthopaedic surgery patients.
Collapse
Affiliation(s)
- Ethan Cottrill
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Julianna Lazzari
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Zach Pennington
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Jeff Ehresman
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Andrew Schilling
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Naomi Dirckx
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Nicholas Theodore
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Daniel Sciubba
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Timothy Witham
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
13
|
Pal S, Porwal K, Rajak S, Sinha RA, Chattopadhyay N. Selective dietary polyphenols induce differentiation of human osteoblasts by adiponectin receptor 1-mediated reprogramming of mitochondrial energy metabolism. Biomed Pharmacother 2020; 127:110207. [PMID: 32422565 DOI: 10.1016/j.biopha.2020.110207] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/18/2020] [Accepted: 04/28/2020] [Indexed: 12/30/2022] Open
Abstract
Anabolic therapies for osteoporosis including dietary polyphenols promote osteoblast function by influencing its energy metabolism. Among the dietary polyphenols, the beneficial skeletal effects of genistein (an isoflavone), kaempferol (a flavone), resveratrol (RES, a stilbenoid) and epigallocatechin gallate (EGCG, a catechin) have been reported in preclinical studies. We studied the action mechanism of these nutraceuticals on osteoblast bioenergetics. All stimulated differentiation of human fetal osteoblasts (hFOB). However, only EGCG and RES stimulated mitochondrial parameters including basal and maximum respiration, spare respiratory capacity and ATP production (a measure of the activity of electron transport chain/ETC). Increases in these parameters were due to increased mitochondrial biogenesis and consequent upregulation of several mitochondrial proteins including those involved in ETC. Rotenone blocked the osteogenic effect of EGCG and RES suggesting the mediatory action of mitochondria. Both compounds rapidly activated AMPK, and dorsomorphin (an AMPK inhibitor) abolished ATP production stimulated by these compounds. Moreover, EGCG and RES upregulated the mitochondrial biogenesis factor, PGC-1α which is downstream of AMPK activation, and silencing PGC-1α blocked their stimulatory effects on ATP production and hFOB differentiation. Adiponectin receptor 1 (AdipoR1) is an upstream regulator of PGC-1α, and both compounds increased the expression of AdipoR1 but not AdipoR2. Silencing AdipoR1 blocked the upregulation of EGCG/RES-induced PGC-1α and hFOB differentiation. In rat calvarium, both compounds increased AdipoR1, PGC-1α, and RunX2 (the osteoblast transcription factor) with a concomitant increase in mitochondrial copy number and ATP levels. We conclude that EGCG and RES display osteogenic effects by reprogramming osteoblastic bioenergetics by acting as the AdipoR1 agonists.
Collapse
Affiliation(s)
- Subhashis Pal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India
| | - Konica Porwal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India
| | - Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Rohit A Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India.
| |
Collapse
|
14
|
Pace JR, Jog R, Burgess DJ, Hadden MK. Formulation and evaluation of itraconazole liposomes for Hedgehog pathway inhibition. J Liposome Res 2019; 30:305-311. [PMID: 31576768 DOI: 10.1080/08982104.2019.1668011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Itraconazole (ITZ) is an FDA-approved antifungal agent that has recently been explored for novel biological properties. In particular, ITZ was identified as a potent inhibitor of the hedgehog (Hh) pathway, a cell signalling pathway that has been linked to a variety of cancers and accounts for ∼25% of paediatric medulloblastoma (MB) cases. To date, there is not a targeted therapeutic option for paediatric MB, resulting in long-term side effects such as hormone deficiency, organ damage and secondary cancers. A primary obstacle for developing targeted therapy for brain ailments is the presence of the blood-brain barrier (BBB), which protects the brain from potentially harmful substances. Due to its size and hydrophobicity, ITZ does not penetrate the BBB. Alternatively, liposomes are being increasingly used within the clinic to increase drug bioavailability, target specificity and BBB permeability. With this in mind, we have successfully developed ITZ-containing liposomes with an optimal size for BBB penetration (<100 nm) and encapsulation efficiency (∼95%) by utilizing a continuous manufacturing approach-turbulent coaxial jet in co-flow. Our preliminary in vitro data demonstrate that these liposomes inhibit the Hh pathway, albeit at a reduced level in comparison to free ITZ. (196/250 words).
Collapse
Affiliation(s)
- Jennifer R Pace
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Rajan Jog
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Diane J Burgess
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - M Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
15
|
Huang Y, Lin Y, Rong M, Liu W, He J, Zhou L. 20(S)-hydroxycholesterol and simvastatin synergistically enhance osteogenic differentiation of marrow stromal cells and bone regeneration by initiation of Raf/MEK/ERK signaling. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:87. [PMID: 31325047 DOI: 10.1007/s10856-019-6284-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 06/24/2019] [Indexed: 06/10/2023]
Abstract
Previous studies have demonstrated the significant roles of simvastatin (SVA) and oxysterols in the osteogenesis process. In this study, we evaluate the effect of a combination of SVA and 20(S)-hydroxycholesterol (20(S)OHC) on the cell viability and osteogenic differentiation of bone marrow stromal cells (BMSCs). After treatment with a control vehicle, SVA (0.025, 0.10, 0.25 or 1.0 μM), 20(S)OHC (5 μM), or a combination of both (0.25 μM SVA + 5 μM 20(S)OHC), the proliferation, apoptosis, ALP activity, mineralization, osteogenesis-related gene expression and Raf/MEK/ERK signaling activity in BMSCs were measured. Our results showed that high concentrations of SVA (0.25 and 1.0 μM) enhanced osteogenesis-related genes expression while attenuating cell viability. The addition of 5 μM 20(S)OHC induced significantly higher proliferative activity, which neutralized the inhibitory effect of SVA on the viability of BMSCs. Moreover, compared to supplementation with only one of the additives, combined supplementation with both SVA and 20(S)OHC induced significantly enhanced ALP activity, calcium sedimentation, osteogenesis-related genes (ALP, OCN and BMP-2) expression and Raf/MEK/ERK signaling activity in BMSCs; these enhancements were attenuated by treatment with the inhibitor U0126, indicating a significant role of Raf/MEK/ERK signaling in mediating the synergistically enhanced osteogenic differentiation of BMSCs by combined SVA and 20(S)OHC treatment. Additionally, histological examination confirmed a synergistic effect of SVA and 20(S)OHC on enhancing bone regeneration in a rabbit calvarial defect model. This newly developed SVA/20(S)OHC formulation may be used as an osteoinductive drug to enhance bone healing.
Collapse
Affiliation(s)
- Yinghe Huang
- Center of Oral Implantology, Stomatological Hospital, Southern Medical University, 366 South Jiangnan Road, Guangzhou, 510280, Guangdong, China
- The Department of Stomatology, Taishan People's Hospital, Affiliated to Guangdong Medical University, Taishan, Guangdong, China
| | - Yao Lin
- The Department of Stomatology, Jieyang Affiliated Hospital, SunYat-sen University, Jieyang, Guangdong, China
| | - Mingdeng Rong
- Center of Oral Implantology, Stomatological Hospital, Southern Medical University, 366 South Jiangnan Road, Guangzhou, 510280, Guangdong, China
| | - Weizhen Liu
- Center of Oral Implantology, Stomatological Hospital, Southern Medical University, 366 South Jiangnan Road, Guangzhou, 510280, Guangdong, China
| | - Junbing He
- The Department of Stomatology, Jieyang Affiliated Hospital, SunYat-sen University, Jieyang, Guangdong, China
| | - Lei Zhou
- Center of Oral Implantology, Stomatological Hospital, Southern Medical University, 366 South Jiangnan Road, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
16
|
Hou W, Ye C, Chen M, Li W, Gao X, He R, Zheng Q, Zhang W. Bergenin Activates SIRT1 as a Novel Therapeutic Agent for Osteogenesis of Bone Mesenchymal Stem Cells. Front Pharmacol 2019; 10:618. [PMID: 31258473 PMCID: PMC6586741 DOI: 10.3389/fphar.2019.00618] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 05/15/2019] [Indexed: 01/18/2023] Open
Abstract
Bone mesenchymal stem cells (BMSCs) are important candidates for bone regeneration. The role of Bergenin, a C-glucoside of 4-O-methyl gallic acid obtained from the species, Bergenia, in BMSC osteogenesis has not yet been elucidated. We therefore investigated the effects of Bergenin on the osteogenesis of BMSCs and found that Bergenin enhanced osteoblast-specific markers and downregulated the adipocyte-specific markers in vitro. Furthermore, using a rat calvarial defect model, we found that Bergenin significantly improved bone healing, as determined by imaging and histological analyses. Moreover, it also upregulated SIRT1 expression. A SIRT1 inhibitor (EX 527) decreased the enhanced bone mineral formation caused by Bergenin. Taken together, these findings show that Bergenin accelerated the osteogenic differentiation of BMSCs, at least partly through the activation of SIRT1.
Collapse
Affiliation(s)
- Weiduo Hou
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Chenyi Ye
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Mo Chen
- Department of Rheumatology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weixu Li
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Xiang Gao
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Rongxin He
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Qiang Zheng
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Wei Zhang
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research institute of Orthopaedics, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Paz JL, Levy D, Oliveira BA, de Melo TC, de Freitas FA, Reichert CO, Rodrigues A, Pereira J, Bydlowski SP. 7-Ketocholesterol Promotes Oxiapoptophagy in Bone Marrow Mesenchymal Stem Cell from Patients with Acute Myeloid Leukemia. Cells 2019; 8:E482. [PMID: 31117185 PMCID: PMC6562391 DOI: 10.3390/cells8050482] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 02/06/2023] Open
Abstract
7-Ketocholesterol (7-KC) is a cholesterol oxidation product with several biological functions. 7-KC has the capacity to cause cell death depending on the concentration and specific cell type. Mesenchymal stem cells (MSCs) are multipotent cells with the ability to differentiate into various types of cells, such as osteoblasts and adipocytes, among others. MSCs contribute to the development of a suitable niche for hematopoietic stem cells, and are involved in the development of diseases, such as leukemia, to a yet unknown extent. Here, we describe the effect of 7-KC on the death of bone marrow MSCs from patients with acute myeloid leukemia (LMSCs). LMSCs were less susceptible to the death-promoting effect of 7-KC than other cell types. 7-KC exposure triggered the extrinsic pathway of apoptosis with an increase in activated caspase-8 and caspase-3 activity. Mechanisms other than caspase-dependent pathways were involved. 7-KC increased ROS generation by LMSCs, which was related to decreased cell viability. 7-KC also led to disruption of the cytoskeleton of LMSCs, increased the number of cells in S phase, and decreased the number of cells in the G1/S transition. Autophagosome accumulation was also observed. 7-KC downregulated the SHh protein in LMSCs but did not change the expression of SMO. In conclusion, oxiapoptophagy (OXIdative stress + APOPTOsis + autophagy) seems to be activated by 7-KC in LMSCs. More studies are needed to better understand the role of 7-KC in the death of LMSCs and the possible effects on the SHh pathway.
Collapse
Affiliation(s)
- Jessica Liliane Paz
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, SP, Brazil.
| | - Debora Levy
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, SP, Brazil.
| | - Beatriz Araujo Oliveira
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, SP, Brazil.
| | - Thatiana Correia de Melo
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, SP, Brazil.
| | - Fabio Alessandro de Freitas
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, SP, Brazil.
| | - Cadiele Oliana Reichert
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, SP, Brazil.
| | - Alessandro Rodrigues
- Departmento de Ciencias Exactas e da Terra, Universidade Federal de Sao Paulo, Diadema 09972-270, SP, Brazil.
| | - Juliana Pereira
- Center of Innovation and Translational Medicine, Department of Medicine, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, SP, Brazil.
| | - Sergio Paulo Bydlowski
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, SP, Brazil.
- Center of Innovation and Translational Medicine, Department of Medicine, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, SP, Brazil.
- National Institute of Science and Technology for Regenerative Medicine (INCT Regenera), CNPq, Rio de Janeiro 21941-902, Brazil.
| |
Collapse
|
18
|
Teng F, Ding H, Huang Y, Wang J. Fabrication of three-dimensional nanofibrous gelatin scaffolds using one-step crosslink technique. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2018; 29:1859-1875. [PMID: 30132379 DOI: 10.1080/09205063.2018.1515299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Electrospun nanofibers have been considered to be an ideal scaffold for tissue engineering, because of the extracellular-matrix-like structure and the well-controlled fabrication. Here, a new method was used to fabricate electrospun three-dimensional macroporous nanofibrous gelatin scaffolds in ethanol bath by one-step crosslink with glutaraldehyde. The mean diameter of the one-step crosslinked fibers was significantly smaller than that of the traditional two-step crosslinked fibers (p < 0.05), and scaffolds prepared by one-step crosslink were fluffy and porous. No significant difference was found in the degradation rates for both fibers within 14 days. After immersion in PBS for 14 days, numerous two-step crosslinked fibers merged together. By contrast, the morphology and macroporous structure of one-step crosslinked fibers showed no evident change and were generally maintained. Approximate crosslinking degrees of the two-step and one-step crosslinked gelatin fibers were 40% and 54%, respectively (p < 0.05). Results from fluorescence microscopy and hematoxylin-eosin staining showed that MC3T3-E1 subclone four cells were distributed more evenly and diversely in the one-step crosslinked fiber scaffolds. The one-step crosslinked fibers enhanced the proliferation and differentiation potential of MC3T3-E1 cells. Furthermore, one-step crosslinked fibers were beneficial in repairing defects in the skulls of rats. Thus, one-step crosslink by glutaraldehyde in ethanol bath is a cost-effective and simple method to fabricate three-dimensional macroporous nanofiberous scaffolds. This technique retains the morphology and structure of the gelatin fibers, and enhances the biological performance of scaffolds in vitro and in vivo.
Collapse
Affiliation(s)
- Fangjun Teng
- a Department of Prosthodontics, Hubei-MOST KLOS & KLOBM , School and Hospital of Stomatology, Wuhan University , Wuhan , China.,b Department of Stomatology, The Central Hospital of Wuhan , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Huifen Ding
- a Department of Prosthodontics, Hubei-MOST KLOS & KLOBM , School and Hospital of Stomatology, Wuhan University , Wuhan , China
| | - Yiqing Huang
- a Department of Prosthodontics, Hubei-MOST KLOS & KLOBM , School and Hospital of Stomatology, Wuhan University , Wuhan , China
| | - Jiawei Wang
- a Department of Prosthodontics, Hubei-MOST KLOS & KLOBM , School and Hospital of Stomatology, Wuhan University , Wuhan , China
| |
Collapse
|
19
|
Shi R, Huang Y, Ma C, Wu C, Tian W. Current advances for bone regeneration based on tissue engineering strategies. Front Med 2018; 13:160-188. [PMID: 30047029 DOI: 10.1007/s11684-018-0629-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/14/2017] [Indexed: 01/07/2023]
Abstract
Bone tissue engineering (BTE) is a rapidly developing strategy for repairing critical-sized bone defects to address the unmet need for bone augmentation and skeletal repair. Effective therapies for bone regeneration primarily require the coordinated combination of innovative scaffolds, seed cells, and biological factors. However, current techniques in bone tissue engineering have not yet reached valid translation into clinical applications because of several limitations, such as weaker osteogenic differentiation, inadequate vascularization of scaffolds, and inefficient growth factor delivery. Therefore, further standardized protocols and innovative measures are required to overcome these shortcomings and facilitate the clinical application of these techniques to enhance bone regeneration. Given the deficiency of comprehensive studies in the development in BTE, our review systematically introduces the new types of biomimetic and bifunctional scaffolds. We describe the cell sources, biology of seed cells, growth factors, vascular development, and the interactions of relevant molecules. Furthermore, we discuss the challenges and perspectives that may propel the direction of future clinical delivery in bone regeneration.
Collapse
Affiliation(s)
- Rui Shi
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Yuelong Huang
- Department of Spine Surgery of Beijing Jishuitan Hospital, The Fourth Clinical Medical College of Peking University, Beijing, 100035, China
| | - Chi Ma
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Chengai Wu
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Wei Tian
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China. .,Department of Spine Surgery of Beijing Jishuitan Hospital, The Fourth Clinical Medical College of Peking University, Beijing, 100035, China.
| |
Collapse
|
20
|
Sulfuretin promotes osteoblastic differentiation in primary cultured osteoblasts and in vivo bone healing. Oncotarget 2018; 7:78320-78330. [PMID: 27713171 PMCID: PMC5346641 DOI: 10.18632/oncotarget.12460] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 09/13/2016] [Indexed: 11/25/2022] Open
Abstract
Although sulfuretin, the major flavonoid of Rhus verniciflua Stokes, has a variety of biological actions, its in vitro and in vivo effects on osteogenic potential remain poorly understood. The objective of the present study was to investigate the effects of sulfuretin on in vitro osteoblastic differentiation and the underlying signal pathway mechanisms in primary cultured osteoblasts and on in vivo bone formation using critical-sized calvarial defects in mice. Sulfuretin promoted osteogenic differentiation of primary osteoblasts, with increased ALP activity and mineralization, and upregulated differentiation markers, including ALP, osteocalcin, and osteopontin, in a concentration-dependent manner. The expression levels of Runx2, BMP-2, and phospho-Smad1/5/8 were upregulated by sulfuretin. Moreover, sulfuretin increased phosphorylation of Akt, mTOR, ERK, and JNK. Furthermore, sulfuretin treatment increased mRNA expression of Wnt ligands, phosphorylation of GSK3, and nuclear β-catenin protein expression. In vivo studies with calvarial bone defects revealed that sulfuretin significantly enhanced new bone formation by micro-computed tomography and histologic analysis. Collectively, these data suggest that sulfuretin acts through the activation of BMP, mTOR, Wnt/β-catenin, and Runx2 signaling to promote in vitro osteoblast differentiation and facilitate in vivo bone regeneration, and might be have therapeutic benefits in bone disease and regeneration.
Collapse
|
21
|
Lee JS, Kim E, Han S, Kang KL, Heo JS. Evaluating the oxysterol combination of 22(S)-hydroxycholesterol and 20(S)-hydroxycholesterol in periodontal regeneration using periodontal ligament stem cells and alveolar bone healing models. Stem Cell Res Ther 2017; 8:276. [PMID: 29208033 PMCID: PMC5717822 DOI: 10.1186/s13287-017-0725-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 11/03/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Oxysterols, oxygenated by-products of cholesterol biosynthesis, play roles in various physiological and pathological systems. However, the effects of oxysterols on periodontal regeneration are unknown. This study investigated the effects of the specific oxysterol combination of 22(S)-hydroxycholesterol and 20(S)-hydroxycholesterol (SS) on the regeneration of periodontal tissues using in-vitro periodontal ligament stem cells (PDLSCs) and in-vivo models of alveolar bone defect. METHODS To evaluate the effects of the combined oxysterols on PDLSC biology, we studied the SS-induced osteogenic differentiation of PDLSCs by assessing alkaline phosphatase activity, intracellular calcium levels [Ca2+]i, matrix mineralization, and osteogenic marker mRNA expression and protein levels. To verify the effect of oxysterols on alveolar bone regeneration, we employed tooth extraction bone defect models. RESULTS Oxysterols increased the osteogenic activity of PDLSCs compared with the control group. The expression of liver X receptor (LXR) α and β, the nuclear receptors for oxysterols, and their target gene, ATP-binding cassette transporter A1 (ABCA1), increased significantly during osteogenesis. Oxysterols also increased protein levels of the hedgehog (Hh) receptor Smo and the transcription factor Gli1. We further confirmed the reciprocal reaction between the LXRs and Hh signaling. Transfection of both LXRα and LXRβ siRNAs decreased Smo and Gli1 protein levels. In contrast, the inhibition of Hh signaling attenuated the LXRα and LXRβ protein levels. Subsequently, SS-induced osteogenic activity of PDLSCs was suppressed by the inhibition of LXRs or Hh signaling. The application of SS also enhanced bone formation in the defect sites of in-vivo models, showing equivalent efficacy to recombinant human bone morphogenetic protein-2. CONCLUSIONS These findings suggest that a specific combination of oxysterols promoted periodontal regeneration by regulating PDLSC activity and alveolar bone regeneration.
Collapse
Affiliation(s)
- Jin-Sun Lee
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul, 02447, South Korea
| | - EunJi Kim
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul, 02447, South Korea
| | - Seonggu Han
- Department of Periodontology, School of Dentistry, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul, 02447, South Korea
| | - Kyung Lhi Kang
- Department of Periodontology, School of Dentistry, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul, 02447, South Korea.
| | - Jung Sun Heo
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul, 02447, South Korea.
| |
Collapse
|
22
|
Levy D, de Melo TC, Ruiz JL, Bydlowski SP. Oxysterols and mesenchymal stem cell biology. Chem Phys Lipids 2017; 207:223-230. [DOI: 10.1016/j.chemphyslip.2017.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/28/2017] [Accepted: 06/28/2017] [Indexed: 02/08/2023]
|
23
|
Silva SF, Levy D, Ruiz JLM, de Melo TC, Isaac C, Fidelis ML, Rodrigues A, Bydlowski SP. Oxysterols in adipose tissue-derived mesenchymal stem cell proliferation and death. J Steroid Biochem Mol Biol 2017; 169:164-175. [PMID: 27133385 DOI: 10.1016/j.jsbmb.2016.04.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/22/2016] [Accepted: 04/27/2016] [Indexed: 11/26/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells characterized by self-renewal and cellular differentiation capabilities. Oxysterols comprise a very heterogeneous group derived from cholesterol through enzymatic and non-enzymatic oxidation. Potent effects in cell death processes, including cytoxicity and apoptosis induction, were described in several cell lines. Very little is known about the effects of oxysterols in MSCs. 7-ketocholesterol (7-KC), one of the most important oxysterols, was shown to be cytotoxic to human adipose tissue-derived MSCs. Here, we describe the short-term (24h) cytotoxic effects of cholestan-3α-5β-6α-triol, 3,5 cholestan-7-one, (3α-5β-6α)- cholestane-3,6-diol, 7-oxocholest-5-en-3β-yl acetate, and 5β-6β epoxy-cholesterol, on MSCs derived from human adipose tissue. MSCs were isolated from adipose tissue obtained from three young, healthy women. Oxysterols, with the exception of 3,5 cholestan-7-one and 7-oxocholest-5-en-3β-yl acetate, led to a complex mode of cell death that include apoptosis, necrosis and autophagy, depending on the type of oxysterol and concentration, being cholestan-3α-5β-6α-triol the most effective. Inhibition of proliferation was also promoted by these oxysterols, but no changes in cell cycle were observed.
Collapse
Affiliation(s)
- Suelen Feitoza Silva
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av.Dr.Enéas de Carvalho Aguiar, 155, 1st floor, room 43, 05403-000, São Paulo/SP, Brazil
| | - Débora Levy
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av.Dr.Enéas de Carvalho Aguiar, 155, 1st floor, room 43, 05403-000, São Paulo/SP, Brazil
| | - Jorge Luis Maria Ruiz
- Federal University of Latin American Integration-UNILA, Life and Nature Science Institute, Av. Tarquinio Joslin dos Santos, 1000, Sala 105, CEP: 85870-901, Foz do Iguacu, Parana/PR, Brazil
| | - Thatiana Correa de Melo
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av.Dr.Enéas de Carvalho Aguiar, 155, 1st floor, room 43, 05403-000, São Paulo/SP, Brazil
| | - Cesar Isaac
- Cell Culture and Wound Healing Research Laboratory, Division of Plastic Surgery, Hospital das Clínicas, University of São Paulo Medical School, Av. Dr. Arnaldo, 455, 1st floor, 05403-000, São Paulo/SP, Brazil
| | - Maíra Luísa Fidelis
- Department of Earth and Exact Sciences, Federal University of São Paulo, São Paulo/SP, Brazil
| | - Alessandro Rodrigues
- Department of Earth and Exact Sciences, Federal University of São Paulo, São Paulo/SP, Brazil
| | - Sérgio Paulo Bydlowski
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av.Dr.Enéas de Carvalho Aguiar, 155, 1st floor, room 43, 05403-000, São Paulo/SP, Brazil.
| |
Collapse
|
24
|
Feasibility of ultra-performance liquid chromatography–ion mobility–time-of-flight mass spectrometry in analyzing oxysterols. J Chromatogr A 2017; 1487:147-152. [DOI: 10.1016/j.chroma.2017.01.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 01/12/2017] [Accepted: 01/15/2017] [Indexed: 12/22/2022]
|
25
|
Kulig W, Cwiklik L, Jurkiewicz P, Rog T, Vattulainen I. Cholesterol oxidation products and their biological importance. Chem Phys Lipids 2016; 199:144-160. [DOI: 10.1016/j.chemphyslip.2016.03.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 12/14/2022]
|
26
|
Maschinot CA, Corman AR, DeBerardinis AM, Hadden MK. Synthesis and Evaluation of Osteogenic Oxysterols as Hedgehog Pathway Activators. ChemMedChem 2016; 11:679-86. [DOI: 10.1002/cmdc.201500550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/12/2016] [Indexed: 01/17/2023]
Affiliation(s)
- Chad A. Maschinot
- Department of Pharmaceutical Sciences; University of Connecticut; 69 North Eagleville Road, Unit 3092 Storrs CT 06269-3092 USA
| | - Audrey R. Corman
- Department of Pharmaceutical Sciences; University of Connecticut; 69 North Eagleville Road, Unit 3092 Storrs CT 06269-3092 USA
| | - Albert M. DeBerardinis
- Department of Pharmaceutical Sciences; University of Connecticut; 69 North Eagleville Road, Unit 3092 Storrs CT 06269-3092 USA
| | - M. Kyle Hadden
- Department of Pharmaceutical Sciences; University of Connecticut; 69 North Eagleville Road, Unit 3092 Storrs CT 06269-3092 USA
| |
Collapse
|
27
|
Balmayor ER. Targeted delivery as key for the success of small osteoinductive molecules. Adv Drug Deliv Rev 2015; 94:13-27. [PMID: 25959428 DOI: 10.1016/j.addr.2015.04.022] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/20/2015] [Accepted: 04/29/2015] [Indexed: 02/08/2023]
Abstract
Molecules such as growth factors, peptides and small molecules can guide cellular behavior and are thus important for tissue engineering. They are rapidly emerging as promising compounds for the regeneration of tissues of the musculoskeletal system. Growth factors have disadvantages such as high cost, short half-life, supraphysiological amounts needed, etc. Therefore, small molecules may be an alternative. These molecules have been discovered using high throughput screening. Small osteoinductive molecules exhibit several advantages over growth factors owing to their small sizes, such as high stability and non-immunogenicity. These molecules may stimulate directly signaling pathways that are important for osteogenesis. However, systemic application doesn't induce osteogenesis in most cases. Therefore, local administration is needed. This may be achieved by using a bone graft material providing additional osteoconductive properties. These graft materials can also act by themselves as a delivery matrix for targeted and local delivery. Furthermore, vascularization is necessary in the process of osteogenesis. Many of the small molecules are also capable of promoting vascularization of the tissue to be regenerated. Thus, in this review, special attention is given to molecules that are capable of inducing both angiogenesis and osteogenesis simultaneously. Finally, more recent preclinical and clinical uses in bone regeneration of those molecules are described, highlighting the needs for the clinical translation of these promising compounds.
Collapse
|
28
|
Wang Y, Zhu G, Li N, Song J, Wang L, Shi X. Small molecules and their controlled release that induce the osteogenic/chondrogenic commitment of stem cells. Biotechnol Adv 2015; 33:1626-40. [PMID: 26341834 DOI: 10.1016/j.biotechadv.2015.08.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/21/2015] [Accepted: 08/23/2015] [Indexed: 12/17/2022]
Abstract
Stem cell-based tissue engineering plays a significant role in skeletal system repair and regenerative therapies. However, stem cells must be differentiated into specific mature cells prior to implantation (direct implantation may lead to tumour formation). Natural or chemically synthesised small molecules provide an efficient, accurate, reversible, and cost-effective way to differentiate stem cells compared with bioactive growth factors and gene-related methods. Thus, investigating the influences of small molecules on the differentiation of stem cells is of great significance. Here, we review a series of small molecules that can induce or/and promote the osteogenic/chondrogenic commitment of stem cells. The controlled release of these small molecules from various vehicles for stem cell-based therapies and tissue engineering applications is also discussed. The extensive studies in this field represent significant contributions to stem cell-based tissue engineering research and regenerative medicine.
Collapse
Affiliation(s)
- Yingjun Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Guanglin Zhu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Nanying Li
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Juqing Song
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Lin Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Xuetao Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China.
| |
Collapse
|
29
|
Li A, Hokugo A, Segovia LA, Yalom A, Rezzadeh K, Zhou S, Zhang Z, Parhami F, Stappenbeck F, Jarrahy R. Oxy133, a novel osteogenic agent, promotes bone regeneration in an intramembranous bone-healing model. J Tissue Eng Regen Med 2015; 11:1490-1499. [PMID: 26073881 DOI: 10.1002/term.2047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 04/10/2015] [Accepted: 04/29/2015] [Indexed: 12/14/2022]
Abstract
Current reconstructive techniques for complex craniofacial osseous defects are challenging and are associated with significant morbidity. Oxysterols are naturally occurring cholesterol oxidation products with osteogenic potential. In this study, we investigated the effects of a novel semi-synthetic oxysterol, Oxy133, on in vitro osteogenesis and an in vivo intramembranous bone-healing model. Rabbit bone marrow stromal cells (BMSCs) were treated with either Oxy133 or BMP-2. Alkaline phosphatase (ALP) activity, expression of osteogenic gene markers and in vitro mineralization were all examined. Next, collagen sponges carrying either Oxy133 or BMP-2 were used to reconstruct critical-sized cranial defects in mature rabbits and bone regeneration was assessed. To determine the mechanism of action of Oxy133 both in vitro and in vivo, rabbit BMSCs cultures and collagen sponge/Oxy133 implants were treated with the Hedgehog signalling pathway inhibitor, cyclopamine, and similar outcomes were measured. ALP activity in rabbit BMSCs treated with 1 μm Oxy133 was induced and was significantly higher than in control cells. These results were mitigated in cultures treated with cyclopamine. Expression of osteogenic gene markers and mineralization in BMSCs treated with 1 μm Oxy133 was significantly higher than in control groups. Complete bone regeneration was noted in vivo when cranial defects were treated with Oxy133; healing was incomplete, however, when cyclopamine was added. Collectively, these results demonstrate that Oxy133 has the ability to induce osteogenic differentiation in vitro in rabbit BMSCs and to promote robust bone regeneration in vivo in an animal model of intramembranous bone healing. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Andrew Li
- Regenerative Bioengineering and Repair Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Akishige Hokugo
- Regenerative Bioengineering and Repair Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Luis Andres Segovia
- Regenerative Bioengineering and Repair Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Anisa Yalom
- Regenerative Bioengineering and Repair Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kameron Rezzadeh
- Regenerative Bioengineering and Repair Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Situo Zhou
- Regenerative Bioengineering and Repair Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Zheyu Zhang
- Regenerative Bioengineering and Repair Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Farhad Parhami
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Reza Jarrahy
- Regenerative Bioengineering and Repair Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
30
|
Kwon IK, Lee SC, Hwang YS, Heo JS. Mitochondrial function contributes to oxysterol-induced osteogenic differentiation in mouse embryonic stem cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:561-72. [DOI: 10.1016/j.bbamcr.2014.12.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/10/2014] [Accepted: 12/10/2014] [Indexed: 02/08/2023]
|
31
|
Mandal CC. High Cholesterol Deteriorates Bone Health: New Insights into Molecular Mechanisms. Front Endocrinol (Lausanne) 2015; 6:165. [PMID: 26557105 PMCID: PMC4617053 DOI: 10.3389/fendo.2015.00165] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 10/08/2015] [Indexed: 01/09/2023] Open
Abstract
Many epidemiological studies show a positive connection between cardiovascular diseases and risk of osteoporosis, suggesting a role of hyperlipidemia and/or hypercholesterolemia in regulating osteoporosis. The majority of the studies indicated a correlation between high cholesterol and high LDL-cholesterol level with low bone mineral density, a strong predictor of osteoporosis. Similarly, bone metastasis is a serious complication of cancer for patients. Several epidemiological and basic studies have established that high cholesterol is associated with increased cancer risk. Moreover, osteoporotic bone environment predisposes the cancer cells for metastatic growth in the bone microenvironment. This review focuses on how cholesterol and cholesterol-lowering drugs (statins) regulate the functions of bone residential osteoblast and osteoclast cells to augment or to prevent bone deterioration. Moreover, this study provides an insight into molecular mechanisms of cholesterol-mediated bone deterioration. It also proposes a potential mechanism by which cellular cholesterol boosts cancer-induced bone metastasis.
Collapse
Affiliation(s)
- Chandi C. Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Rajasthan, India
- *Correspondence: Chandi C. Mandal,
| |
Collapse
|
32
|
Zarrouk A, Vejux A, Mackrill J, O’Callaghan Y, Hammami M, O’Brien N, Lizard G. Involvement of oxysterols in age-related diseases and ageing processes. Ageing Res Rev 2014; 18:148-62. [PMID: 25305550 DOI: 10.1016/j.arr.2014.09.006] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/23/2014] [Accepted: 09/30/2014] [Indexed: 12/15/2022]
Abstract
Ageing is accompanied by increasing vulnerability to major pathologies (atherosclerosis, Alzheimer's disease, age-related macular degeneration, cataract, and osteoporosis) which can have similar underlying pathoetiologies. All of these diseases involve oxidative stress, inflammation and/or cell death processes, which are triggered by cholesterol oxide derivatives, also named oxysterols. These oxidized lipids result either from spontaneous and/or enzymatic oxidation of cholesterol on the steroid nucleus or on the side chain. The ability of oxysterols to induce severe dysfunctions in organelles (especially mitochondria) plays key roles in RedOx homeostasis, inflammatory status, lipid metabolism, and in the control of cell death induction, which may at least in part contribute to explain the potential participation of these molecules in ageing processes and in age related diseases. As no efficient treatments are currently available for most of these diseases, which are predicted to become more prevalent due to the increasing life expectancy and average age, a better knowledge of the biological activities of the different oxysterols is of interest, and constitutes an important step toward identification of pharmacological targets for the development of new therapeutic strategies.
Collapse
|
33
|
Lee JS, Yang JH, Hong JY, Jung UW, Yang HC, Lee IS, Choi SH. Early bone healing onto implant surface treated by fibronectin/oxysterol for cell adhesion/osteogenic differentiation: in vivo experimental study in dogs. J Periodontal Implant Sci 2014; 44:242-50. [PMID: 25368813 PMCID: PMC4216401 DOI: 10.5051/jpis.2014.44.5.242] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 09/28/2014] [Indexed: 01/17/2023] Open
Affiliation(s)
- Jung-Seok Lee
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
| | - Jin-Hyuk Yang
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
| | - Ji-Youn Hong
- Department of Periodontology, Kyung Hee University School of Dentistry, Seoul, Korea
| | - Ui-Won Jung
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
| | - Hyeong-Cheol Yang
- Department of Dental Biomaterials Science, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - In-Seop Lee
- Atomic-Scale Surface Science Research Center, Yonsei University, Seoul, Korea
| | - Seong-Ho Choi
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
| |
Collapse
|
34
|
Resveratrol improves bone repair by modulation of bone morphogenetic proteins and osteopontin gene expression in rats. Int J Oral Maxillofac Surg 2014; 43:900-6. [DOI: 10.1016/j.ijom.2014.01.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 12/02/2013] [Accepted: 01/13/2014] [Indexed: 12/28/2022]
|
35
|
Levy D, Ruiz JLM, Celestino AT, Silva SF, Ferreira AK, Isaac C, Bydlowski SP. Short-term effects of 7-ketocholesterol on human adipose tissue mesenchymal stem cells in vitro. Biochem Biophys Res Commun 2014; 446:720-5. [PMID: 24491549 DOI: 10.1016/j.bbrc.2014.01.132] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 01/25/2014] [Indexed: 01/27/2023]
Abstract
Oxysterols comprise a very heterogeneous group derived from cholesterol through enzymatic and non-enzymatic oxidation. Among them, 7-ketocholesterol (7-KC) is one of the most important. It has potent effects in cell death processes, including cytoxicity and apoptosis induction. Mesenchymal stem cells (MSCs) are multipotent cells characterized by self-renewal and cellular differentiation capabilities. Very little is known about the effects of oxysterols in MSCs. Here, we describe the short-term cytotoxic effect of 7-ketocholesterol on MSCs derived from human adipose tissue. MSCs were isolated from adipose tissue obtained from two young, healthy women. After 24 h incubation with 7-KC, mitochondrial hyperpolarization was observed, followed by a slight increase in the level of apoptosis and changes in actin organization. Finally, the IC50 of 7-KC was higher in these cells than has been observed or described in other normal or cancer cell lines.
Collapse
Affiliation(s)
- Débora Levy
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av. Dr. Enéas de Carvalho Aguiar,155, 1st Floor, Room 43, 05403-000 São Paulo, SP, Brazil
| | - Jorge Luis Maria Ruiz
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av. Dr. Enéas de Carvalho Aguiar,155, 1st Floor, Room 43, 05403-000 São Paulo, SP, Brazil
| | - Andrea Turbuck Celestino
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av. Dr. Enéas de Carvalho Aguiar,155, 1st Floor, Room 43, 05403-000 São Paulo, SP, Brazil
| | - Suelen Feitoza Silva
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av. Dr. Enéas de Carvalho Aguiar,155, 1st Floor, Room 43, 05403-000 São Paulo, SP, Brazil
| | - Adilson Kleber Ferreira
- Departament of Immunology, Laboratory of Tumor Immunology, Institute of Biomedical Science, University of Sao Paulo, Av. Prof. Lineu Prestes, 1730-Room 254, 05508-900 São Paulo, SP, Brazil
| | - Cesar Isaac
- Cell Culture and Wound Healing Research Laboratory, Division of Plastic Surgery, University of São Paulo, Av. Dr. Arnaldo, 455, 1st Floor, 05403-000 São Paulo, SP, Brazil
| | - Sérgio Paulo Bydlowski
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av. Dr. Enéas de Carvalho Aguiar,155, 1st Floor, Room 43, 05403-000 São Paulo, SP, Brazil.
| |
Collapse
|
36
|
Abstract
AIMS To investigate the effect of 1-(4-(tert-butyl)benzyl)-N-(4-methoxyphenyl)-3-phenyl-1H-pyrazole-5-carboxamide (Pyr-C) on the proliferation and osteogenic differentiation of MC3T3-E1 cells. MATERIALS & METHODS MTT and BrdU incorporation assay were used to determine cell survival and proliferation. The gene expression levels of osteogenic markers were determined using real-time PCR and ALP activity was detected. Western-blot analysis was used to determine the protein expression of BSP and OPN. The long-term effect of Pyr-C on mineralization deposition was measured by Alizarin Red Staining. RESULTS Pyr-C inhibited cell proliferation and increased ALP activity. Gene expression of ALP, BSP, OCN, Runx2, and Osterix was up-regulated in Pyr-C-induced group. Pyr-C increased the protein expression of BSP at day 7, 14 and 21, and OPN at day 14, 21 and 28. Meanwhile, Pyr-C enhanced the mineral deposition. CONCLUSION Pyr-C inhibits proliferation and stimulates osteogenic differentiation of MC3T3-E1 cells.
Collapse
|
37
|
Hokugo A, Sorice S, Parhami F, Yalom A, Li A, Zuk P, Jarrahy R. A novel oxysterol promotes bone regeneration in rabbit cranial bone defects. J Tissue Eng Regen Med 2013; 10:591-9. [PMID: 23997014 DOI: 10.1002/term.1799] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 06/20/2013] [Accepted: 07/15/2013] [Indexed: 12/16/2022]
Abstract
Bone morphogenetic proteins (BMPs) have played a central role in the development of regenerative therapies for bone reconstruction. However, the high cost and side-effect profile of BMPs limits their broad application. Oxysterols, naturally occurring products of cholesterol oxidation, are promising osteogenic agents alternative to BMPs. The osteogenic capacity of these non-toxic and relatively inexpensive molecules has been documented in rodent models. We studied the impact of Oxy49, a novel oxysterol analogue, on the osteogenic differentiation of rabbit bone marrow stromal cells (BMSCs). Moreover, we evaluated the capacity for in vivo bone regeneration with Oxy49 in rabbit cranial bone defects. We found that rabbit BMSCs treated with Oxy49 demonstrated differentiation along osteogenic pathways, and that complete bone regeneration occurred when cranial defects were treated with Oxy49. Collectively, these results demonstrate that Oxy49 has the ability to induce osteogenic differentiation in rabbit BMSCs with an efficacy comparable to that of BMP-2 and to promote significant bone regeneration in cranial defects. Oxysterols may be a viable novel agent in bone tissue engineering. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Akishige Hokugo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sarah Sorice
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Farhad Parhami
- Department of Medicine, David Geffen School of Medicine at UCLA, Center for the Health Sciences, Los Angeles, CA, USA
| | - Anisa Yalom
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Andrew Li
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Patricia Zuk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Reza Jarrahy
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
38
|
Lo KWH, Ashe KM, Kan HM, Laurencin CT. The role of small molecules in musculoskeletal regeneration. Regen Med 2013; 7:535-49. [PMID: 22817627 DOI: 10.2217/rme.12.33] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The uses of bone morphogenetic proteins and parathyroid hormone therapeutics are fraught with several fundamental problems, such as cost, protein stability, immunogenicity, contamination and supraphysiological dosage. These downsides may effectively limit their more universal use. Therefore, there is a clear need for alternative forms of biofactors to obviate the drawbacks of protein-based inductive factors for bone repair and regeneration. Our group has studied small molecules with the capacity to regulate osteoblast differentiation and mineralization because their inherent physical properties minimize limitations observed in protein growth factors. For instance, in general, small molecule inducers are usually more stable, highly soluble, nonimmunogenic, more affordable and require lower dosages. Small molecules with the ability to induce osteoblastic differentiation may represent the next generation of bone regenerative medicine. This review describes efforts to develop small molecule-based biofactors for induction, paying specific attention to their novel roles in bone regeneration.
Collapse
Affiliation(s)
- Kevin W-H Lo
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA
| | | | | | | |
Collapse
|
39
|
Corman A, DeBerardinis AM, Hadden MK. Structure-activity relationships for side chain oxysterol agonists of the hedgehog signaling pathway. ACS Med Chem Lett 2012; 3:828-33. [PMID: 24900386 DOI: 10.1021/ml300192k] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 08/28/2012] [Indexed: 01/20/2023] Open
Abstract
Oxysterols (OHCs) are byproducts of cholesterol oxidation that are known to activate the Hedeghog (Hh) signaling pathway. While OHCs that incorporate hydroxyl groups throughout the scaffold are known, those that act as agonists of Hh signaling primarily contain a single hydroxyl on the alkyl side chain. We sought to further explore how side chain hydroxylation patterns affect oxysterol-mediated Hh activation, by performing a structure-activity relationship study on a series of synthetic OHCs. The most active analogue, 23(R)-OHC (35), demonstrated potent activation of Hh signaling in two Hh-dependent cell lines (EC50 values 0.54-0.65 μM). In addition, OHC 35 was approximately 3-fold selective for the Hh pathway as compared to the liver X receptor, a nuclear receptor that is also activated by endogenous OHCs. Finally, 35 induced osteogenic differentiation and osteoblast formation in cultured cells, indicating functional agonism of the Hh pathway.
Collapse
Affiliation(s)
- Audrey Corman
- Department
of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269-3092,
United States
| | - Albert M. DeBerardinis
- Department
of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269-3092,
United States
| | - M. Kyle Hadden
- Department
of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269-3092,
United States
| |
Collapse
|
40
|
Johnson JS, Meliton V, Kim WK, Lee KB, Wang JC, Nguyen K, Yoo D, Jung ME, Atti E, Tetradis S, Pereira RC, Magyar C, Nargizyan T, Hahn TJ, Farouz F, Thies S, Parhami F. Novel oxysterols have pro-osteogenic and anti-adipogenic effects in vitro and induce spinal fusion in vivo. J Cell Biochem 2011; 112:1673-84. [PMID: 21503957 DOI: 10.1002/jcb.23082] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Stimulation of bone formation by osteoinductive materials is of great clinical importance in spinal fusion surgery, repair of bone fractures, and in the treatment of osteoporosis. We previously reported that specific naturally occurring oxysterols including 20(S)-hydroxycholesterol (20S) induce the osteogenic differentiation of pluripotent mesenchymal cells, while inhibiting their adipogenic differentiation. Here we report the characterization of two structural analogues of 20S, Oxy34 and Oxy49, which induce the osteogenic and inhibit the adipogenic differentiation of bone marrow stromal cells (MSC) through activation of Hedgehog (Hh) signaling. Treatment of M2-10B4 MSC with Oxy34 or Oxy49 induced the expression of osteogenic differentiation markers Runx2, Osterix (Osx), alkaline phosphatase (ALP), bone sialoprotein (BSP), and osteocalcin (OCN), as well as ALP enzymatic activity and robust mineralization. Treatment with oxysterols together with PPARγ activator, troglitazone (Tro), inhibited mRNA expression for adipogenic genes PPARγ, LPL, and aP2, and inhibited the formation of adipocytes. Efficacy of Oxy34 and Oxy49 in stimulating bone formation in vivo was assessed using the posterolateral intertransverse process rat spinal fusion model. Rats receiving collagen implants with Oxy 34 or Oxy49 showed comparable osteogenic efficacy to BMP2/collagen implants as measured by radiography, MicroCT, and manual inspection. Histological analysis showed trabecular and cortical bone formation by oxysterols and rhBMP2 within the fusion mass, with robust adipogenesis in BMP2-induced bone and significantly less adipocytes in oxysterol-induced bone. These data suggest that Oxy34 and Oxy49 are effective novel osteoinductive molecules and may be suitable candidates for further development and use in orthopedic indications requiring local bone formation.
Collapse
Affiliation(s)
- Jared S Johnson
- Department of Orthopedic Surgery, UCLA, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Carvalho JFS, Silva MMC, Moreira JN, Simões S, Sá e Melo ML. Selective Cytotoxicity of Oxysterols through Structural Modulation on Rings A and B. Synthesis, in Vitro Evaluation, and SAR. J Med Chem 2011; 54:6375-93. [DOI: 10.1021/jm200803d] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- João F. S. Carvalho
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - M. Manuel Cruz Silva
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculdade de Farmácia, Universidade de Coimbra, 3000-548 Coimbra, Portugal
| | - João N. Moreira
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculdade de Farmácia, Universidade de Coimbra, 3000-548 Coimbra, Portugal
| | - Sérgio Simões
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculdade de Farmácia, Universidade de Coimbra, 3000-548 Coimbra, Portugal
| | - M. Luisa Sá e Melo
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculdade de Farmácia, Universidade de Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
42
|
Yang Y, Hallgrimsson B, Putnins EE. Craniofacial defect regeneration using engineered bone marrow mesenchymal stromal cells. J Biomed Mater Res A 2011; 99:74-85. [PMID: 21800417 DOI: 10.1002/jbm.a.33155] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 04/12/2011] [Accepted: 04/26/2011] [Indexed: 12/24/2022]
Abstract
Large craniofacial bony defects remain a significant clinical challenge. Bone marrow mesenchymal stromal cells (BM-MSCs) constitute a multipotent population. Previously, we developed a novel approach for BM-MSC expansion on 3D CultiSpher-S gelatin microcarrier beads in spin culture with preservation of their multipotentiality, reduction of apoptosis, and enhancement of bone formation in vivo. Here, we hypothesized that such cultured BM-MSCs without exogenous growth factors would respond to the orthopedic microenvironment, thus promoting craniofacial defect regeneration. BM-MSCs isolated from green fluorescent protein (GFP) transgenic rats were ex vivo expanded and transplanted into critical-sized (5-mm diameter) rat calvaria defects. Gelatin beads or defect alone served as controls. By 28 and 42 days, rats were sacrificed for microcomputed tomography (microCT), histologic, and immunohistochemistry examination. MicroCT results demonstrated that BM-MSCs were a statistically significant factor contributing to new bone volume regeneration. Histologic assessment showed that the BM-MSCs group produced more and higher quality new bone compared with beads or defect-alone groups in both osteoinductive and osteoconductive manners. Specifically, immunohistochemical staining identified GFP(+) cells residing in new bone lacunae in conjunction with non-GFP(+) cells. Therefore, ex vivo expanded BM-MSCs at least in part regenerated critical-sized calvaria defects by osteogenic differentiation in vivo.
Collapse
Affiliation(s)
- Yi Yang
- Faculty of Dentistry, The University of British Columbia, Vancouver, BC, Canada V6T1Z3
| | | | | |
Collapse
|
43
|
Mackrill JJ. Oxysterols and calcium signal transduction. Chem Phys Lipids 2011; 164:488-95. [PMID: 21513705 DOI: 10.1016/j.chemphyslip.2011.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 04/04/2011] [Accepted: 04/06/2011] [Indexed: 12/31/2022]
Abstract
Ionised calcium (Ca(2+)) is a key second messenger, regulating almost every cellular process from cell death to muscle contraction. Cytosolic levels of this ion can be increased via gating of channel proteins located in the plasma membrane, endoplasmic reticulum and other membrane-delimited organelles. Ca(2+) can be removed from cells by extrusion across the plasma membrane, uptake into organelles and buffering by anionic components. Ca(2+) channels and extrusion mechanisms work in concert to generate diverse spatiotemporal patterns of this second messenger, the distinct profiles of which determine different cellular outcomes. Increases in cytoplasmic Ca(2+) concentration are one of the most rapid cellular responses upon exposure to certain oxysterol congeners or to oxidised low-density lipoprotein, occurring within seconds of addition and preceding increases in levels of reactive oxygen species, or changes in gene expression. Furthermore, exposure of cells to oxysterols for periods of hours to days modulates Ca(2+) signal transduction, with these longer-term alterations in cellular Ca(2+) homeostasis potentially underlying pathological events within atherosclerotic lesions, such as hyporeactivity to vasoconstrictors observed in vascular smooth muscle, or ER stress-induced cell death in macrophages. Despite their candidate roles in physiology and disease, little is known about the molecular mechanisms that couple changes in oxysterol concentrations to alterations in Ca(2+) signalling. This review examines the ways in which oxysterols could influence Ca(2+) signal transduction and the potential roles of this in health and disease.
Collapse
Affiliation(s)
- John J Mackrill
- Department of Physiology, University College Cork, Cork, Ireland.
| |
Collapse
|
44
|
Son KM, Park HC, Kim NR, Lee IS, Yang HC. Enhancement of the ALP activity of C3H10T1/2 cells by the combination of an oxysterol and apatite. Biomed Mater 2010; 5:044107. [DOI: 10.1088/1748-6041/5/4/044107] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
45
|
Abstract
We previously reported that specific oxysterols stimulate osteogenic differentiation of pluripotent bone marrow stromal cells (MSCs) through activation of hedgehog (Hh) signaling and may serve as potential future therapies for intervention in osteopenia and osteoporosis. In this study we report that the osteogenic oxysterol 20(S)-hydroxycholesterol (20S) induces the expression of genes associated with Notch signaling. Using M2-10B4 (M2) MSCs, we found that 20S significantly induced HES-1, HEY-1, and HEY-2 mRNA expression compared with untreated cells, with maximal induction after 48 hours, whereas the nonosteogenic oxysterols did not. Similar observations were made when M2 cells were treated with sonic hedgehog (Shh), and the specific Hh pathway inhibitor cyclopamine blocked 20S-induced Notch target gene expression. 20S did not induce Notch target genes in Smo(-/-) mouse embryonic fibroblasts, further confirming the role of Hh signaling in 20S-induced expression of Notch target genes. Despite the inability of liver X-receptor (LXR) synthetic ligand TO901317 to induce Notch target genes in M2 cells, LXR knockdown studies using siRNA showed inhibition of 20S-induced HEY-1 but not HES-1 expression, suggesting the partial role of LXR signaling in MSC responses to 20S. Moreover, 20S-induced Notch target gene expression was independent of canonical Notch signaling because neither 20S nor Shh induced CBF1 luciferase reporter activity or NICD protein accumulation in the nucleus, which are hallmarks of canonical Notch signaling activation. Finally, HES-1 and HEY-1 siRNA transfection significantly inhibited 20S-induced osteogenic genes, suggesting that the pro-osteogenic effects of 20S are regulated in part by HES-1 and HEY-1.
Collapse
|
46
|
Plaisant M, Fontaine C, Cousin W, Rochet N, Dani C, Peraldi P. Activation of hedgehog signaling inhibits osteoblast differentiation of human mesenchymal stem cells. Stem Cells 2009; 27:703-13. [PMID: 19096040 DOI: 10.1634/stemcells.2008-0888] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells within the bone are responsible for the generation of osteoblasts, chondrocytes, and adipocytes. In rodents, Indian hedgehog has been shown to play a role in osteoblast differentiation. However, evidence for a direct function of hedgehog (Hh) in human osteoblastic differentiation is missing. Using different models of human mesenchymal stem cells we show that Hh signaling decreases during osteoblast differentiation. This is associated with a decrease in Smoothened expression, a key partner that triggers Hh signaling, and in the number of cells displaying a primary cilium, an organelle necessary for Hh signaling. Remarkably, treatment of human mesenchymal stem cells with sonic hedgehog or two molecules able to activate Hh signaling inhibits osteoblast differentiation. This inhibition is visualized through a decrease in mineralization and in the expression of osteoblastic genes. In particular, activation of Hh signaling induces a decrease in Runx2 expression, a key transcriptional factor controlling the early stage of osteoblast differentiation. Consistently, the activation of Hh signaling during the first days of differentiation is sufficient to inhibit osteoblast differentiation, whereas differentiated osteoblasts are not affected by Hh signaling. In summary, we show here, using various inducers of Hh signaling and mesenchymal stem cells of two different origins, that Hh signaling inhibits human osteoblast differentiation, in sharp contrast to what has been described in rodent cells. This species difference should be taken into account for screening for pro-osteogenic molecules.
Collapse
Affiliation(s)
- Magali Plaisant
- Institute of Signaling, Biology, Development and Cancer, Université de Nice Sophia-Antipolis, CNRS UMR, France
| | | | | | | | | | | |
Collapse
|
47
|
The small molecule phenamil induces osteoblast differentiation and mineralization. Mol Cell Biol 2009; 29:3905-14. [PMID: 19433444 DOI: 10.1128/mcb.00002-09] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stimulation of osteoblast differentiation from mesenchymal stem cells is a potential strategy for bone repair. Bone morphogenetic proteins (BMPs) that induce osteoblastic differentiation have been successfully used in humans to treat fractures. Here we outline a new approach to the stimulation of osteoblast differentiation using small molecules that stimulate BMP activity. We have identified the amiloride derivative phenamil as a stimulator of osteoblast differentiation and mineralization. Remarkably, phenamil acts cooperatively with BMPs to induce the expression of BMP target genes, osteogenic markers, and matrix mineralization in both mesenchymal stem cell lines and calvarial organ cultures. Transcriptional profiling of cells treated with phenamil led to the identification of tribbles homolog 3 (Trb3) as a mediator of its effects. Trb3 is induced by phenamil selectively in cells with osteoblastic potential. Both Trb3 and phenamil stabilize the expression of SMAD, the critical transcription factor in BMP signaling, by promoting the degradation of SMAD ubiquitin regulatory factor 1. Small interfering RNA-mediated knockdown of Trb3 blunts the effects of phenamil on BMP signaling and osteogenesis. Thus, phenamil induces osteogenic differentiation, at least in part, through Trb3-dependent promotion of BMP action. The synergistic use of small molecules such as phenamil along with BMPs may provide new strategies for the promotion of bone healing.
Collapse
|
48
|
Umoh JU, Sampaio AV, Welch I, Pitelka V, Goldberg HA, Underhill TM, Holdsworth DW. In vivomicro-CT analysis of bone remodeling in a rat calvarial defect model. Phys Med Biol 2009; 54:2147-61. [DOI: 10.1088/0031-9155/54/7/020] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
49
|
Magyar CE, Aghaloo TL, Atti E, Tetradis S. Ostene, a new alkylene oxide copolymer bone hemostatic material, does not inhibit bone healing. Neurosurgery 2008; 63:373-8; discussion 378. [PMID: 18981846 DOI: 10.1227/01.neu.0000316859.03788.44] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE In this study, we investigate the effects of a soft bone hemostatic wax comprised of water-soluble alkylene oxide copolymers (Ostene; Ceremed, Inc., Los Angeles, CA) on bone healing in a rat calvaria defect model. We compared the effects with a control (no hemostatic agent) and bone wax, an insoluble and nonresorbable material commonly used for bone hemostasis. METHODS Two bilateral 3-mm circular noncritical-sized defects were made in the calvariae of 30 rats. Alkylene oxide copolymer or bone wax was applied or no hemostatic material was used (control). After 3, 6, and 12 weeks, rats were sacrificed and the calvariae excised. Bone healing, expressed as fractional bone volume (+/- standard error of the mean), was measured by microcomputed tomography. RESULTS Immediate hemostasis was achieved equally with bone wax and alkylene oxide copolymer. Bone wax-filled defects remained unchanged at all time points with negligible healing observed. At 3 weeks, no evidence of alkylene oxide copolymer was observed at the application site, with fractional bone volume significantly greater than bone wax-treated defects (0.20 +/- 0.03 versus 0.02 +/- 0.01; P = 0.0003). At 6 and 12-weeks, alkylene oxide copolymer-treated defects continued to show significantly greater healing versus bone wax (0.18 +/- 0.04 versus 0.05 +/- 0.01 and 0.31 +/- 0.04 versus 0.06 +/- 0.02, respectively). At all time points, alkylene oxide copolymer-treated and control defects showed good healing with no significant difference. CONCLUSION Alkylene oxide copolymer is an effective hemostatic agent that does not inhibit osteogenesis or bone healing.
Collapse
Affiliation(s)
- Clara E Magyar
- Department of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, California 90095, USA.
| | | | | | | |
Collapse
|
50
|
Ramanadham S, Yarasheski KE, Silva MJ, Wohltmann M, Novack DV, Christiansen B, Tu X, Zhang S, Lei X, Turk J. Age-related changes in bone morphology are accelerated in group VIA phospholipase A2 (iPLA2beta)-null mice. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:868-81. [PMID: 18349124 DOI: 10.2353/ajpath.2008.070756] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Phospholipases A(2) (PLA(2)) hydrolyze the sn-2 fatty acid substituent, such as arachidonic acid, from phospholipids, and arachidonate metabolites are recognized mediators of bone modeling. We have previously generated knockout (KO) mice lacking the group VIA PLA(2) (iPLA(2)beta), which participates in a variety of signaling events; iPLA(2)beta mRNA is expressed in bones of wild-type (WT) but not KO mice. Cortical bone size, trabecular bone volume, bone mineralizing surfaces, and bone strength are similar in WT and KO mice at 3 months and decline with age in both groups, but the decreases are more pronounced in KO mice. The lower bone mass phenotype observed in KO mice is not associated with an increase in osteoclast abundance/activity or a decrease in osteoblast density, but is accompanied by an increase in bone marrow fat. Relative to WT mice, undifferentiated bone marrow stromal cells (BMSCs) from KO mice express higher levels of PPAR-gamma and lower levels of Runx2 mRNA, and this correlates with increased adipogenesis and decreased osteogenesis in BMSCs from these mice. In summary, our studies indicate that age-related losses in bone mass and strength are accelerated in iPLA(2)beta-null mice. Because adipocytes and osteoblasts share a common mesenchymal stem cell origin, our findings suggest that absence of iPLA(2)beta causes abnormalities in osteoblast function and BMSC differentiation and identify a previously unrecognized role of iPLA(2)beta in bone formation.
Collapse
Affiliation(s)
- Sasanka Ramanadham
- Washington University School of Medicine, Department of Internal Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|